Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Redox Biol ; 67: 102898, 2023 11.
Article in English | MEDLINE | ID: mdl-37757542

ABSTRACT

TNFα-mediated signaling pathways play a pivotal role in the pathogenesis of inflammatory diseases such as rheumatoid arthritis (RA) and inflammatory bowel disease (IBD) by promoting phagocyte inflammatory functions, notably cytokine release and reactive oxygen species (ROS) production by NOX2. In contrast, interleukin-10 (IL-10), a powerful anti-inflammatory cytokine, potently shuts down phagocyte activation, making IL-10 an attractive therapeutic candidate. However, IL-10 therapy has shown limited efficacy in patients with inflammatory diseases. Here, we report that TNFα blocks IL-10 anti-inflammatory pathways in human monocytes, thereby prolonging inflammation. TNFα decreased IL-10-induced phosphorylation of STAT3 and consequently IL-10-induced expression of the major anti-inflammatory factor, SOCS3. Decreased STAT3 phosphorylation was due to a SHP1/2 phosphatase, as NSC-87877, a SHP1/2 inhibitor, restored STAT3 phosphorylation and prevented the TNFα-induced inhibition of IL-10 signaling. TNFα activated only SHP1 in human monocytes and this activation was NOX2-dependent, as diphenyleneiodonium, a NOX2 inhibitor, suppressed SHP1 activation and STAT3 dephosphorylation triggered by TNFα. ROS-induced activation of SHP1 was mediated by the redox-sensitive kinase, Lyn, as its inhibition impeded TNFα-induced SHP1 activation and STAT3 dephosphorylation. Furthermore, H2O2 recapitulated TNFα-inhibitory activity on IL-10 signaling. Finally, NSC-87877 dampened collagen antibody-induced arthritis (CAIA) in mice. These results reveal that TNFα disrupts IL-10 signaling by inducing STAT3 dephosphorylation through a NOX2-ROS-Lyn-SHP1 axis in human monocytes and that inhibition of SHP1/2 in vivo protects against CAIA. These new findings might explain the poor efficacy of IL-10 therapy in patients with inflammatory diseases and suggest that anti-TNFα agents and SHP1/2 inhibitors could improve the therapeutic use of IL-10.


Subject(s)
Interleukin-10 , Monocytes , Humans , Animals , Mice , Monocytes/metabolism , Interleukin-10/genetics , Interleukin-10/metabolism , Tumor Necrosis Factor-alpha/metabolism , Reactive Oxygen Species/metabolism , Hydrogen Peroxide/metabolism , Cytokines/metabolism , Anti-Inflammatory Agents , STAT3 Transcription Factor/metabolism
2.
Proc Natl Acad Sci U S A ; 120(3): e2209184120, 2023 01 17.
Article in English | MEDLINE | ID: mdl-36626553

ABSTRACT

Monocytes play a key role in innate immunity by eliminating pathogens, releasing high levels of cytokines, and differentiating into several cell types, including macrophages and dendritic cells. Similar to other phagocytes, monocytes produce superoxide anions through the NADPH oxidase complex, which is composed of two membrane proteins (p22phox and gp91phox/NOX2) and four cytosolic proteins (p47phox, p67phox, p40phox and Rac1). The pathways involved in NADPH oxidase activation in monocytes are less known than those in neutrophils. Here, we show that p22phox is associated with Rho-associated coiled-coil kinase 2 (ROCK2) in human monocytes but not neutrophils. This interaction occurs between the cytosolic region of p22phox (amino acids 132 to 195) and the coiled-coil region of ROCK2 (amino acids 400 to 967). Interestingly, ROCK2 does not phosphorylate p22phox, p40phox, p67phox, or gp91phox in vitro but phosphorylates p47phox on Ser304, Ser315, Ser320 and Ser328. Furthermore, KD025, a selective inhibitor of ROCK2, inhibited reactive oxygen species (ROS) production and p47phox phosphorylation in monocytes. Specific inhibition of ROCK2 expression in THP1-monocytic cell line by siRNA inhibited ROS production. These data show that ROCK2 interacts with p22phox and phosphorylates p47phox, and suggest that p22phox could be a shuttle for ROCK2 to allow p47phox phosphorylation and NADPH oxidase activation in human monocytes.


Subject(s)
Monocytes , NADPH Oxidases , rho-Associated Kinases , Humans , Amino Acids , Monocytes/metabolism , NADPH Oxidases/metabolism , Phosphoproteins/metabolism , Reactive Oxygen Species , rho-Associated Kinases/metabolism
3.
Blood ; 139(16): 2512-2522, 2022 04 21.
Article in English | MEDLINE | ID: mdl-35108370

ABSTRACT

Superoxide production by the phagocyte reduced NAD phosphate (NADPH) oxidase is essential for innate immunity as shown in chronic granulomatous disease (CGD), an immunodeficiency disease resulting from mutations in 1 of its genes. The NADPH oxidase is composed of 2 membrane proteins (gp91phox/NOX2 and p22phox) and 4 cytosolic proteins (p47phox, p67phox, p40phox, and Rac1/2). The phosphorylation of p47phox is required for NADPH oxidase activation in cells. As p47phox and p67phox can form a tight complex in cells, we hypothesized that p67phox could regulate p47phox phosphorylation. To investigate this hypothesis, we used phospho-specific antibodies against 5 major p47phox-phosphorylated sites (Ser304, Ser315, Ser320, Ser328, and Ser345) and neutrophils from healthy donors and from p67phox-/- CGD patients. Results showed that formyl-methionyl-leucyl-phenylalanine and phorbol myristate acetate induced a time- and a concentration-dependent phosphorylation of p47phox on Ser304, Ser315, Ser320, and Ser328 in healthy human neutrophils. Interestingly, in neutrophils and Epstein-Barr virus-transformed B lymphocytes from p67phox-/- CGD patients, phosphorylation of p47phox on serine residues was dramatically reduced. In COSphox cells, the presence of p67phox led to increased phosphorylation of p47phox. In vitro studies showed that recombinant p47phox was phosphorylated on Ser304, Ser315, Ser320, and Ser328 by different PKC isoforms and the addition of recombinant p67phox alone or in combination with p40phox potentiated this process. Thus, p67phox and p40phox are required for optimal p47phox phosphorylation on Ser304, Ser315, Ser320, and Ser328 in intact cells. Therefore, p67phox and p40phox are novel regulators of p47phox-phosphorylation.


Subject(s)
Epstein-Barr Virus Infections , Granulomatous Disease, Chronic , Enzyme Activation , Epstein-Barr Virus Infections/metabolism , Granulomatous Disease, Chronic/genetics , Herpesvirus 4, Human/metabolism , Humans , NADPH Oxidases/genetics , NADPH Oxidases/metabolism , Neutrophils/metabolism , Phosphoproteins/genetics , Phosphoproteins/metabolism , Phosphorylation
4.
Cell Mol Gastroenterol Hepatol ; 13(4): 1073-1093, 2022.
Article in English | MEDLINE | ID: mdl-35031518

ABSTRACT

BACKGROUND & AIMS: NADPH oxidase 1 (NOX1) has emerged as a prime regulator of intestinal mucosa immunity and homeostasis. Dysregulation of NOX1 may cause inflammatory bowel disease (IBD). It is not clear how NOX1 is regulated in vivo under inflammatory conditions. We studied the role of CK2 in this process. METHODS: The NOX1 organizer subunit, NADPH oxidase organizer 1 (NOXO1), was immunoprecipitated from cytokine-treated colon epithelial cells, and bound proteins were identified by mass spectrometry analysis. Sites on NOXO1 phosphorylated by CK2 were identified by nanoscale liquid chromatography coupled to tandem mass spectrometry. NOX1 activity was determined in colon epithelial cells and colonoids in the presence or absence of CX-4945, a CK2 specific inhibitor. Acute colitis was induced by administration of trinitrobenzenesulfonic acid in mice treated or not with CX-4945. Colon tissues were analyzed by histologic examination, quantitative polymerase chain reaction, and Western blots. CK2 activity, markers of inflammation, and oxidative stress were assessed. RESULTS: We identified CK2 as a major partner of NOXO1 in colon epithelial cells under inflammatory conditions. CK2 directly binds NOXO1 at the C-terminus containing the Phox homology domain and phosphorylates NOXO1 on several sites. CX-4945 increased ROS generation by NOX1 in human colon epithelial cells and organoids. Strikingly, CK2 activity was reduced in trinitrobenzenesulfonic acid-induced acute colitis, and CX-4945 exacerbated colitis inflammation as shown by increased levels of CXCL1, ROS generation, lipid peroxidation, and colon damage. CONCLUSIONS: The ubiquitous protein kinase CK2 limits NOX1 activity via NOXO1 binding and phosphorylation in colonic epithelial cells and lessens experimental colitis. Loss of CK2 activity during acute colitis results in excessive ROS production, contributing to the pathogenesis. Strategies to activate CK2 could be an effective novel therapeutic approach in IBD.


Subject(s)
Colitis , Inflammatory Bowel Diseases , Animals , Casein Kinase II/adverse effects , Colitis/chemically induced , Inflammation , Mice , NADPH Oxidase 1/metabolism , Reactive Oxygen Species/metabolism , Trinitrobenzenesulfonic Acid/adverse effects
5.
Biomedicines ; 9(9)2021 Sep 01.
Article in English | MEDLINE | ID: mdl-34572316

ABSTRACT

Neutrophils are key cells of the innate immune and inflammatory responses. They are the first blood cells to migrate to the infection site where they release high amounts of reactive oxygen species (ROS) and several peptides and enzymes required for microbial killing. However, excessive neutrophil activation can induce tissue injury participating in inflammation, thus the characterization of the enzymes involved in neutrophil activation could help to identify new pharmacological targets to treat inflammation. The prolyl-isomerase Pin1 is a ubiquitous enzyme involved in several functions, however, its role in neutrophil functions is less known. In this study, we show that the bacterial peptide N-formyl-methionyl-leucyl-phenylalanine (fMLP or fMLF), a G-protein coupled receptor (GPCR) agonist-induced Pin1 activation in human neutrophils. PiB and juglone, two Pin1 inhibitors inhibited Pin1 activity in neutrophils and consequently inhibited fMLP-induced chemotaxis and -degranulation of azurophil and specific granules as measured by myeloperoxidase and neutrophil gelatinase-associated lipocalin (NGAL) release respectively. We also showed that PiB inhibited TNFα + fMLP-induced superoxide production, confirming the effect of juglone. These data show that inhibitors of Pin1 impaired key pro-inflammatory neutrophil functions elicited by GPCR activation and suggest that Pin1 could control neutrophil inflammatory functions.

7.
Biochem Pharmacol ; 177: 113950, 2020 07.
Article in English | MEDLINE | ID: mdl-32251677

ABSTRACT

Neutrophils are key cells in innate immunity and inflammation. Granulocyte-macrophage colony-stimulating factor (GM-CSF) is known to enhance many neutrophil functions such as reactive oxygen species (ROS) production, degranulation and cell survival via the activation of the ERK1/2 pathway. ERK1/2 pathway activation is redox sensitive and could be modulated by ROS. In order to investigate whether NADPH oxidase NOX2-derived ROS could contribute to GM-CSF-induced ERK1/2 phosphorylation, we tested the effect of two selective NOX2 inhibitors, diphenylene iodonium (DPI) and apocynin. Results showed that, while both DPI and apocynin strongly inhibited neutrophil ROS production, only apocynin, but not DPI, inhibited GM-CSF-induced ERK1/2 phosphorylation, suggesting that ROS are not involved in this process. Apocynin did not affect GM-CSF-induced p38MAPKinase phosphorylation, another redox sensitive kinase. Interestingly, apocynin inhibited GM-CSF-induced MEK1/2 and AKT phosphorylation without affecting fMLF-induced phosphorylation of these proteins. GM-CSF is known to inhibit neutrophils apoptosis and to promote cell survival via the AKT-ERK1/2 pathway. In this regard, we found that apocynin also inhibited GM-CSF-induced anti-apoptotic effect in neutrophils. These results suggest that NADPH oxidase NOX2-derived ROS are not involved in GM-CSF-induced ERK1/2 phosphorylation and that apocynin inhibits GM-CSF-induced ERK1/2 phosphorylation pathway independently of its inhibitory action on NADPH oxidase NOX2. Thus, apocynin can exert an anti-inflammatory effect not only by limiting neutrophil ROS production but also by decreasing neutrophil survival at inflammatory site.


Subject(s)
Acetophenones/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , NADPH Oxidases/metabolism , Neutrophils/drug effects , Phagocytes/drug effects , Antioxidants/pharmacology , Apoptosis/drug effects , Cell Survival/drug effects , Cells, Cultured , Enzyme Activation/drug effects , Humans , Neutrophils/cytology , Phagocytes/metabolism , Phosphorylation/drug effects , Reactive Oxygen Species/metabolism
8.
Inflammopharmacology ; 28(2): 487-497, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31667656

ABSTRACT

Excessive reactive oxygen species (ROS) production can induce tissue injury involved in a variety of neurodegenerative disorders such as neurodegeneration observed in pilocarpine-induced temporal lobe epilepsy. Ketamine, a noncompetitive N-methyl-D-aspartate (NMDA) receptor antagonist has beneficial effects in pilocarpine-induced temporal lobe epilepsy, when administered within minutes of seizure to avoid the harmful neurological lesions induced by pilocarpine. However, the enzymes involved in ROS productions and the effect of ketamine on this process remain less documented. Here we show that during pilocarpine-induced epilepsy in mice, the expression of the phagocyte NADPH oxidase NOX2 subunits (NOX2/gp91phox, p22phox, and p47phox) and the expression of myeloperoxidase (MPO) were dramatically increased in mice brain treated with pilocarpine. Interestingly, treatment of mice with ketamine before or after pilocarpine administration decreased this process, mainly when injected before pilocarpine. Finally, our results showed that pilocarpine induced p47phox phosphorylation and H2O2 production in mice brain and ketamine was able to inhibit these processes. Our results show that pilocarpine induced NOX2 activation to produce ROS in mice brain and that administration of ketamine before or after the induction of temporal lobe epilepsy by pilocarpine inhibited this activation in mice brain. These results suggest a key role of the phagocyte NADPH oxidase NOX2 and MPO in epilepsy and identify a novel effect of ketamine.


Subject(s)
Epilepsy, Temporal Lobe/drug therapy , Excitatory Amino Acid Antagonists/pharmacology , Ketamine/pharmacology , Reactive Oxygen Species/metabolism , Animals , Brain/physiopathology , Disease Models, Animal , Epilepsy, Temporal Lobe/physiopathology , Mice , NADPH Oxidase 2/metabolism , NADPH Oxidases/metabolism , Peroxidase/metabolism , Phagocytes/metabolism , Phosphorylation , Pilocarpine
9.
Sci Rep ; 9(1): 18540, 2019 12 06.
Article in English | MEDLINE | ID: mdl-31811262

ABSTRACT

Eugenol is a polyphenol extracted from Syzygium aromaticum essential oil. It is known to have anti-inflammatory and chemoprotective properties as well as a potent anti-oxidant activity due the presence of its phenolic group. In this study, we examined the effects of eugenol on neutrophil superoxide production, a key process involved in innate immunity and inflammation. Superoxide anion generationin human neutrophils was measured by cytochrome c reduction assay. Western blotting was used to analyze the phosphorylation of, p47phox, MAPKinases (p38 and ERK1/2), MEK1/2 and Raf, key proteins involved in the activation of NADPH oxidase. Pretreatment of neutrophils by increasing concentrations (2.5 µg/mL-20 µg/mL) of eugenol for 30 min, inhibited significantly (p < 0.001) superoxide anion generation induced by the chemotactic peptide formyl-Met-Leu-Phe (fMLF) with an IC50 of 5 µg/mL. Phorbolmyristate acetate (PMA)-stimulated O2- production was affected only at the highest eugenol concentration (20 µg/mL). Results showed that eugenol decreased the phosphorylation of p47phox onSer-345 and Ser-328, the translocation of p47phox to the membranesand the phosphorylation of Raf, MEK1/2 and ERK1/2 proteins. Taken together, our results suggest that eugenol inhibits the generation of superoxide anion by neutrophils via the inhibition of Raf/MEK/ERK1/2/p47phox-phosphorylation pathway.


Subject(s)
Antioxidants/pharmacology , Eugenol/pharmacology , MAP Kinase Signaling System/drug effects , NADPH Oxidases/metabolism , Neutrophils/drug effects , Chemotaxis/immunology , Healthy Volunteers , Humans , MAP Kinase Signaling System/immunology , N-Formylmethionine Leucyl-Phenylalanine/metabolism , NADPH Oxidases/antagonists & inhibitors , Neutrophils/immunology , Neutrophils/metabolism , Oxidative Stress/drug effects , Oxidative Stress/immunology , Phosphorylation/drug effects , Primary Cell Culture , Superoxides/metabolism
10.
Front Immunol ; 10: 2567, 2019.
Article in English | MEDLINE | ID: mdl-31736979

ABSTRACT

Production of superoxide anion and other reactive oxygen species (ROS) by neutrophils has a vital role in host defense against microbes. However, over-production can induce cell injury participating to inflammation. Superoxide anion is produced by the phagocyte NADPH oxidase/NOX2, a multicomponent enzyme system consisting of six proteins: two trans-membrane proteins (gp91 phox and p22 phox ) and four soluble cytosolic proteins (p40 phox , p67 phox , p47 phox , and the small G-proteins, Rac1/2). Phosphorylation of p47 phox on several serines regulates NADPH oxidase activation. LPS released by gram negative bacteria can enhance or prime neutrophil superoxide production in combination with other agonists such as the bacterial peptide formyl-Met-Leu-Phe (fMLP). Since the pathways involved in LPS-induced priming are not completely understood, we investigated the role of the prolyl cis/trans isomerase Pin1 in this process. Two different Pin1 inhibitors, PiB, and Juglone are able to block LPS-induced priming of ROS production by human neutrophils in a concentration dependent manner. PiB and Juglone did not inhibit LPS-induced CD11b translocation neither CD62L shedding. LPS induced an increase of Pin1 activity in neutrophils similar to TNFα and fMLP. Since the phosphorylation of p47 phox on Ser345 is critical for NADPH oxidase up-regulation, we investigated the effect of LPS on this process. Results show that LPS induced the phosphorylation of p47 phox mainly on serine 345 and induced the activation of p38MAPKinase and ERK1/2. These results suggest that the prolyl cis/trans isomerase Pin1 may control LPS-induced priming of superoxide production in human neutrophils. Pharmacological targeting of Pin1 could be a valuable approach in sepsis.


Subject(s)
NADPH Oxidase 2/immunology , NIMA-Interacting Peptidylprolyl Isomerase/immunology , Neutrophils/immunology , Humans , Lipopolysaccharides , Mitogen-Activated Protein Kinases/immunology , N-Formylmethionine Leucyl-Phenylalanine , NIMA-Interacting Peptidylprolyl Isomerase/antagonists & inhibitors , Naphthoquinones/pharmacology , Neutrophils/drug effects , Reactive Oxygen Species/immunology , Sepsis/immunology
12.
J Exp Med ; 216(11): 2669-2687, 2019 11 04.
Article in English | MEDLINE | ID: mdl-31492810

ABSTRACT

Neutrophils produce high levels of reactive oxygen species (ROS) by NADPH oxidase that are crucial for host defense but can lead to tissue injury when produced in excess. We previously described that proliferating cell nuclear antigen (PCNA), a nuclear scaffolding protein pivotal in DNA synthesis, controls neutrophil survival through its cytosolic association with procaspases. We herein showed that PCNA associated with p47phox, a key subunit of NADPH oxidase, and that this association regulated ROS production. Surface plasmon resonance and crystallography techniques demonstrated that the interdomain-connecting loop of PCNA interacted directly with the phox homology (PX) domain of the p47phox. PCNA inhibition by competing peptides or by T2AA, a small-molecule PCNA inhibitor, decreased NADPH oxidase activation in vitro. Furthermore, T2AA provided a therapeutic benefit in mice during trinitro-benzene-sulfonic acid (TNBS)-induced colitis by decreasing oxidative stress, accelerating mucosal repair, and promoting the resolution of inflammation. Our data suggest that targeting PCNA in inflammatory neutrophils holds promise as a multifaceted antiinflammatory strategy.


Subject(s)
Cytosol/metabolism , NADPH Oxidase 2/metabolism , NADPH Oxidases/metabolism , Neutrophils/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , Colitis/chemically induced , Colitis/prevention & control , Enzyme Activation/drug effects , Female , Humans , Male , Mice, Inbred C57BL , Mice, Knockout , NADPH Oxidases/genetics , Protein Binding , Reactive Oxygen Species/metabolism , Small Molecule Libraries/pharmacology , Trinitrobenzenesulfonic Acid
13.
J Immunol ; 202(5): 1549-1558, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30665935

ABSTRACT

Superoxide anion production by the phagocyte NADPH oxidase plays a crucial role in host defenses and inflammatory reaction. The phagocyte NADPH oxidase is composed of cytosolic components (p40phox, p47phox, p67phox, and Rac1/2) and the membrane flavocytochrome b558, which is composed of two proteins: p22phox and gp91phox/NOX2. p22phox plays a crucial role in the stabilization of gp91phox in phagocytes and is also a docking site for p47phox during activation. In the current study, we have used a yeast two-hybrid approach to identify unknown partners of p22phox. Using the cytosolic C-terminal region of p22phox as bait to screen a human spleen cDNA library, we identified the protein interacting with amyloid precursor protein tail 1 (PAT1) as a potential partner of p22phox. The interaction between p22phox and PAT1 was further confirmed by in vitro GST pulldown and overlay assays and in intact neutrophils and COSphox cells by coimmunoprecipitation. We demonstrated that PAT1 is expressed in human neutrophils and monocytes and colocalizes with p22phox, as shown by confocal microscopy. Overexpression of PAT1 in human monocytes and in COSphox cells increased superoxide anion production and depletion of PAT1 by specific small interfering RNA inhibited this process. These data clearly identify PAT1 as a novel regulator of NADPH oxidase activation and superoxide anion production, a key phagocyte function.


Subject(s)
Amino Acid Transport Systems/metabolism , Phagocytes/metabolism , Superoxides/metabolism , Symporters/metabolism , Amino Acid Transport Systems/genetics , Anions/metabolism , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , Symporters/genetics
14.
Mucosal Immunol ; 12(1): 117-131, 2019 01.
Article in English | MEDLINE | ID: mdl-30279516

ABSTRACT

Inflammatory bowel disease (IBD) is characterized by severe and recurrent inflammation of the gastrointestinal tract, associated with altered patterns of cytokine synthesis, excessive reactive oxygen species (ROS) production, and high levels of the innate immune protein, lipocalin-2 (LCN-2), in the mucosa. The major source of ROS in intestinal epithelial cells is the NADPH oxidase NOX1, which consists of the transmembrane proteins, NOX1 and p22PHOX, and the cytosolic proteins, NOXO1, NOXA1, and Rac1. Here, we investigated whether NOX1 activation and ROS production induced by key inflammatory cytokines in IBD causally affects LCN-2 production in colonic epithelial cells. We found that the combination of TNFα and IL-17 induced a dramatic upregulation of NOXO1 expression that was dependent on the activation of p38MAPK and JNK1/2, and resulted into an increase of NOX1 activity and ROS production. NOX1-derived ROS drive the expression of LCN-2 by controlling the expression of IκBζ, a master inducer of LCN-2. Furthermore, LCN-2 production and colon damage were decreased in NOX1-deficient mice during TNBS-induced colitis. Finally, analyses of biopsies from patients with Crohn's disease showed increased JNK1/2 activation, and NOXO1 and LCN-2 expression. Therefore, NOX1 might play a key role in mucosal immunity and inflammation by controlling LCN-2 expression.


Subject(s)
Colitis/immunology , Colon/metabolism , Inflammatory Bowel Diseases/immunology , Intestinal Mucosa/metabolism , Lipocalin-2/metabolism , NADPH Oxidase 1/metabolism , Reactive Oxygen Species/metabolism , Animals , Cells, Cultured , Colitis/chemically induced , Colon/pathology , Cytochrome b Group/genetics , Cytokines/metabolism , Gene Expression Regulation , Humans , Immunity, Innate , Interleukin-17/metabolism , Intestinal Mucosa/pathology , Lipocalin-2/genetics , Mice , Mice, Knockout , NADPH Oxidase 1/genetics , NADPH Oxidases/genetics , RNA, Small Interfering/genetics , Signal Transduction , Trinitrobenzenesulfonic Acid/toxicity , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
15.
Blood ; 130(15): 1734-1745, 2017 10 12.
Article in English | MEDLINE | ID: mdl-28830888

ABSTRACT

Dendritic cells (DCs) are a heterogeneous population of professional antigen-presenting cells and are key cells of the immune system, acquiring different phenotypes in accordance with their localization during the immune response. A subset of inflammatory DCs is derived from circulating monocytes (Mo) and has a key role in inflammation and infection. The pathways controlling Mo-DC differentiation are not fully understood. Our objective was to investigate the possible role of nicotinamide adenine dinucleotide phosphate reduced form oxidases (NOXs) in Mo-DC differentiation. In this study, we revealed that Mo-DC differentiation was inhibited by NOX inhibitors and reactive oxygen species scavengers. We show that the Mo-DC differentiation was dependent on p22phox, and not on gp91phox/NOX2, as shown by the reduced Mo-DC differentiation observed in chronic granulomatous disease patients lacking p22phox. Moreover, we revealed that NOX5 expression was strongly increased during Mo-DC differentiation, but not during Mo-macrophage differentiation. NOX5 was expressed in circulating myeloid DC, and at a lower level in plasmacytoid DC. Interestingly, NOX5 was localized at the outer membrane of the mitochondria and interacted with p22phox in Mo-DC. Selective inhibitors and small interfering RNAs for NOX5 indicated that NOX5 controlled Mo-DC differentiation by regulating the JAK/STAT/MAPK and NFκB pathways. These data demonstrate that the NOX5-p22phox complex drives Mo-DC differentiation, and thus could be critical for immunity and inflammation.


Subject(s)
Cell Differentiation , Dendritic Cells/cytology , Membrane Proteins/metabolism , Monocytes/cytology , NADPH Oxidases/metabolism , Cell Differentiation/drug effects , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Enzyme Inhibitors/pharmacology , Free Radical Scavengers/pharmacology , Humans , Membrane Glycoproteins/metabolism , Membrane Proteins/antagonists & inhibitors , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Membranes/drug effects , Mitochondrial Membranes/metabolism , Models, Biological , Monocytes/drug effects , Monocytes/metabolism , NADPH Oxidase 2 , NADPH Oxidase 5 , NADPH Oxidases/antagonists & inhibitors , NF-kappa B/metabolism , Protein Binding/drug effects , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects
16.
Mol Ecol ; 26(7): 1802-1817, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27661780

ABSTRACT

Although microbial ecology of the gut is now a major focus of interest, little is known about the molecular determinants of microbial adaptation in the gut. Experimental evolution coupled with whole-genome sequencing can provide insights of the adaptive process. In vitro experiments have revealed some conserved patterns: intermediate convergence, and epistatic interactions between beneficial mutations and mutations in global regulators. To test the relevance of these patterns and to identify the selective pressures acting in vivo, we have performed a long-term adaptation of an E. coli natural isolate, the streptomycin-resistant strain 536, in the digestive tract of streptomycin-treated mice. After a year of evolution, a clone from 15 replicates was sequenced. Consistently with in vitro observations, the identified mutations revealed a strong pattern of convergence at the mutation, gene, operon and functional levels. Yet, the rate of molecular evolution was lower than in in vitro, and no mutations in global regulators were recovered. More specific targets were observed: the dgo operon, involved in the galactonate pathway that improved growth on D-galactonate, and rluD and gidB, implicated in the maturation of the ribosomes, which mutations improved growth only in the presence of streptomycin. As in vitro, the nonrandom associations of mutations within the same pathways suggested a role of epistasis in shaping the adaptive landscape. Overall, we show that 'evolve and sequence' approach coupled with an analysis of convergence, when applied to a natural isolate, can be used to study adaptation in vivo and uncover the specific selective pressures of that environment.


Subject(s)
Adaptation, Physiological , Escherichia coli/genetics , Evolution, Molecular , Gastrointestinal Tract/microbiology , Streptomycin/pharmacology , Animals , Drug Resistance, Bacterial , Escherichia coli/drug effects , Female , Genes, Bacterial , Mice , Mutation , Operon
17.
PLoS One ; 9(9): e108738, 2014.
Article in English | MEDLINE | ID: mdl-25268639

ABSTRACT

The Escherichia coli species is divided in phylogenetic groups that differ in their virulence and commensal distribution. Strains belonging to the B2 group are involved in extra-intestinal pathologies but also appear to be more prevalent as commensals among human occidental populations. To investigate the genetic specificities of B2 sub-group, we used 128 sequenced genomes and identified genes of the core genome that showed marked difference between B2 and non-B2 genomes. We focused on the gene and its surrounding region with the strongest divergence between B2 and non-B2, the antiporter gene nhaA. This gene is part of the nhaAR operon, which is in the core genome but flanked by mobile regions, and is involved in growth at high pH and high sodium concentrations. Consistently, we found that a panel of non-B2 strains grew faster than B2 at high pH and high sodium concentrations. However, we could not identify differences in expression of the nhaAR operon using fluorescence reporter plasmids. Furthermore, the operon deletion had no differential impact between B2 and non-B2 strains, and did not result in a fitness modification in a murine model of gut colonization. Nevertheless, sequence analysis and experiments in a murine model of septicemia revealed that recombination in nhaA among B2 strains was observed in strains with low virulence. Finally, nhaA and nhaAR operon deletions drastically decreased virulence in one B2 strain. This effect of nhaAR deletion appeared to be stronger than deletion of all pathogenicity islands. Thus, a population genetic approach allowed us to identify an operon in the core genome without strong effect in commensalism but with an important role in extra-intestinal virulence, a landmark of the B2 strains.


Subject(s)
DNA-Binding Proteins/genetics , Escherichia coli Proteins/genetics , Escherichia coli/pathogenicity , Sodium-Hydrogen Exchangers/genetics , Transcription Factors/genetics , Animals , DNA-Binding Proteins/classification , DNA-Binding Proteins/metabolism , Disease Models, Animal , Escherichia coli/growth & development , Escherichia coli Proteins/classification , Escherichia coli Proteins/metabolism , Female , Genome, Bacterial , Hydrogen-Ion Concentration , Mice , Operon , Osmolar Concentration , Phenotype , Phylogeny , Sepsis/microbiology , Sepsis/mortality , Sepsis/pathology , Sodium-Hydrogen Exchangers/classification , Sodium-Hydrogen Exchangers/metabolism , Survival Rate , Transcription Factors/classification , Transcription Factors/metabolism , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL